Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Chem Biol Interact ; 392: 110905, 2024 Apr 01.
Article in English | MEDLINE | ID: mdl-38373627

ABSTRACT

Aldose reductase is a member of the 1B1 subfamily of aldo-keto reductase gene superfamily. The action of aldose reductase (AR) has been implicated in the pathogenesis of a variety of disease states, most notably complications of diabetes mellitus including neuropathy, retinopathy, nephropathy, and cataracts. To explore for mechanistic roles for AR in disease pathogenesis, we established mutant strains produced using Crispr-Cas9 to inactivate the AKR1B3 gene in C57BL6 mice. Phenotyping AR-knock out (ARKO) strains confirmed previous reports of reduced accumulation of tissue sorbitol levels. Lens epithelial cells in ARKO mice showed markedly reduced epithelial-to-mesenchymal transition following lens extraction in a surgical model of cataract and posterior capsule opacification. A previously unreported phenotype of preputial sebaceous gland swelling was observed frequently in male ARKO mice homozygous for the mutant AKR1B3 allele. This condition, which was shown to be accompanied by infiltration of proinflammatory CD3+ lymphocytes, was not observed in WT mice or mice heterozygous for the mutant allele. Despite this condition, reproductive fitness of the ARKO strain was indistinguishable from WT mice housed under identical conditions. These studies establish the utility of a new strain of AKR1B3-null mice created to support mechanistic studies of cataract and diabetic eye disease.


Subject(s)
Capsule Opacification , Cataract , Lens, Crystalline , Animals , Male , Mice , Aldehyde Reductase/genetics , Capsule Opacification/pathology , Cataract/genetics , Cataract/pathology , Incidence , Inflammation/pathology , Lens, Crystalline/pathology , Mice, Inbred C57BL , Mice, Knockout , Sebaceous Glands
2.
Nutrients ; 15(20)2023 Oct 16.
Article in English | MEDLINE | ID: mdl-37892451

ABSTRACT

Excessive intake of sugar, and particularly fructose, is closely associated with the development and progression of metabolic syndrome in humans and animal models. However, genetic disorders in fructose metabolism have very different consequences. While the deficiency of fructokinase, the first enzyme involved in fructose metabolism, is benign and somewhat desirable, missense mutations in the second enzyme, aldolase B, causes a very dramatic and sometimes lethal condition known as hereditary fructose intolerance (HFI). To date, there is no cure for HFI, and treatment is limited to avoiding fructose and sugar. Because of this, for subjects with HFI, glucose is their sole source of carbohydrates in the diet. However, clinical symptoms still occur, suggesting that either low amounts of fructose are still being consumed or, alternatively, fructose is being produced endogenously in the body. Here, we demonstrate that as a consequence of consuming high glycemic foods, the polyol pathway, a metabolic route in which fructose is produced from glucose, is activated, triggering a deleterious mechanism whereby glucose, sorbitol and alcohol induce severe liver disease and growth retardation in aldolase B knockout mice. We show that generically and pharmacologically blocking this pathway significantly improves metabolic dysfunction and thriving and increases the tolerance of aldolase B knockout mice to dietary triggers of endogenous fructose production.


Subject(s)
Digestive System Diseases , Fructose Intolerance , Liver Diseases , Humans , Animals , Mice , Fructose Intolerance/genetics , Fructose Intolerance/diagnosis , Fructose/metabolism , Fructose-Bisphosphate Aldolase/genetics , Glucose/therapeutic use , Mice, Knockout
3.
Retin Cases Brief Rep ; 16(4): 515-519, 2022 Jul 01.
Article in English | MEDLINE | ID: mdl-32541433

ABSTRACT

PURPOSE: To present a case of idiopathic retinal vasculitis, aneurysms, and neuroretinitis syndrome that was successfully managed with serial intravitreal aflibercept injections. METHODS: Ophthalmic imaging and visual acuity were used to monitor disease state and track treatment methods to determine the most valuable combination of treatment medication and treatment interval. RESULTS: A 28-year-old woman with idiopathic retinal vasculitis, aneurysms, and neuroretinitis syndrome status after panretinal photocoagulation of both eyes presented with bilateral cystoid macular edema. We demonstrate successful management of retinal cystoid macular edema associated with idiopathic retinal vasculitis, aneurysms, and neuroretinitis syndrome using serial intravitreal aflibercept injections. CONCLUSION: Intravitreal aflibercept has a useful role in managing the potential retinal complications associated with idiopathic retinal vasculitis, aneurysms, and neuroretinitis syndrome and provides further insights into treatment of the later stages of this rare disease.


Subject(s)
Aneurysm , Macular Edema , Retinal Vasculitis , Retinitis , Adult , Aneurysm/therapy , Angiogenesis Inhibitors/therapeutic use , Dementia , Female , Hearing Loss, Central , Humans , Intravitreal Injections , Macular Edema/diagnosis , Macular Edema/drug therapy , Macular Edema/etiology , Optic Atrophy , Receptors, Vascular Endothelial Growth Factor , Recombinant Fusion Proteins , Retinal Vasculitis/complications , Retinal Vasculitis/diagnosis , Retinal Vasculitis/drug therapy , Retinitis/diagnosis , Retinitis/drug therapy
4.
PLoS One ; 16(12): e0248545, 2021.
Article in English | MEDLINE | ID: mdl-34971554

ABSTRACT

INTRODUCTION: Cerebrospinal fluid (CSF) outflow has been demonstrated along nasal lymphatics via olfactory nerve projections; flow may be increased by stimulating lymphatic contractility using agents such as noradrenaline and the thromboxane A2 analog U46619. Lymphatics elsewhere in the body show increased contractility upon exposure to the prostaglandin F2alpha analog isoprostane-8-epi-prostaglandin. We investigated the ability of ophthalmic prostaglandin F2alpha analogs to increase CSF outflow when applied to the nasal mucosa by inhalation. METHODS: Latanoprost (0.1, 0.5, or 1mg/ml), bimatoprost (0.3 or 3mg/ml), travoprost (0.04 or 0.4mg/ml), latanoprostene bunod (0.24 or 2.4mg/ml), tafluprost (0.25 or 2.5mg/ml), or control vehicle (10% DMSO) was administered to awake adult C57B/6 mice by nasal inhalation of 2µl droplets. Multiday dosing (daily for 3 days) of latanoprost also was evaluated. A total of 81 animals were studied including controls. General anesthesia was induced by injection, and fluorescent tracer (AlexaFluor647-labelled ovalbumin) was injected under stereotaxic guidance into the right lateral ventricle. Nasal turbinate tissue was harvested and homogenized after 1 hour for tracer detection by ELISA and fluorometric analysis. RESULTS: Inhalation of latanoprost 0.5mg/ml and 1mg/ml led to a 11.5-fold increase in tracer recovery from nasal turbinate tissues compared to controls (3312 pg/ml vs 288 pg/ml, p<0.001 for 0.5mg/ml; 3355 pg/ml vs 288 pg/ml, p<0.001 for 1mg/ml), while latanoprost 0.1 mg/ml enhanced recovery 6-fold (1713 pg/ml vs 288 pg/ml, p<0.01). Tafluprost 0.25mg/ml and bimatoprost 0.3mg/ml showed a modest (1.4x, p<0.05) effect, and the remaining agents showed no significant effect on tracer recovery. After 3 days of daily latanoprost treatment and several hours after the last dose, a persistently increased recovery of tracer was found. CONCLUSIONS: Prostaglandin F2alpha analogs delivered by nasal inhalation resulted in increased nasal recovery of a CSF fluorescent tracer, implying increased CSF outflow via the nasal lymphatics. The greatest effect, partially dose-dependent, was observed using latanoprost. Further studies are needed to determine the efficacy of these agents in reducing ICP in short and long-term applications.


Subject(s)
Absorption, Physiological , Cerebrospinal Fluid/metabolism , Nasal Mucosa/metabolism , Prostaglandins, Synthetic/pharmacology , Absorption, Physiological/drug effects , Administration, Intranasal , Animals , Dinoprost/analogs & derivatives , Female , Fluorescent Dyes/chemistry , Fluorometry , Latanoprost , Male , Mice, Inbred C57BL , Nasal Mucosa/drug effects
5.
Biomolecules ; 11(8)2021 08 04.
Article in English | MEDLINE | ID: mdl-34439816

ABSTRACT

Cataracts are a leading cause of blindness worldwide. Surgical removal of cataracts is a safe and effective procedure to restore vision. However, a large number of patients later develop vision loss due to regrowth of lens cells and subsequent degradation of the visual axis leading to visual disability. This postsurgical complication, known as posterior capsular opacification (PCO), occurs in up to 30% of cataract patients and has no clinically proven pharmacological means of prevention. Despite the availability of many compounds capable of preventing early steps in PCO development, there is currently no effective means to deliver such therapies into the eye for a suitable duration. To model a solution to this unmet medical need, we fabricated acrylic substrates as intraocular lens (IOL) mimics scaled to place into the capsular bag of the mouse lens following a mock-cataract surgery. Substrates were coated with a hydrophilic crosslinked acrylate nanogel designed to elute Sorbinil, an aldose reductase inhibitor previously shown to suppress PCO. Insertion of the Sorbinil-eluting device into the lens capsule at the time of cataract surgery resulted in substantial prevention of cellular changes associated with PCO development. This model demonstrates that a cataract inhibitor can be delivered into the postsurgical lens capsule at therapeutic levels.


Subject(s)
Capsule Opacification/prevention & control , Cataract Extraction/adverse effects , Drug Carriers , Enzyme Inhibitors/pharmacology , Imidazolidines/pharmacology , Lenses, Intraocular , Actins/genetics , Actins/metabolism , Animals , Cadherins/genetics , Cadherins/metabolism , Capsule Opacification/etiology , Capsule Opacification/genetics , Capsule Opacification/pathology , Cataract/genetics , Cataract/metabolism , Cataract/pathology , Cataract Extraction/methods , Disease Models, Animal , Fibronectins/genetics , Fibronectins/metabolism , Gene Expression Regulation , Humans , Lens, Crystalline/metabolism , Lens, Crystalline/pathology , Lens, Crystalline/surgery , Mice , Nanogels/administration & dosage , Nanogels/chemistry , Signal Transduction , Vimentin/genetics , Vimentin/metabolism
6.
Invest Ophthalmol Vis Sci ; 62(10): 24, 2021 08 02.
Article in English | MEDLINE | ID: mdl-34415985

ABSTRACT

Purpose: To determine the effect of metformin on early Nd:YAG laser treatment for posterior capsule opacification (PCO) and to explore a molecular mechanism to explain a possible protective effect of metformin against PCO. Methods: We conducted: 1) a retrospective cohort study of patient eyes undergoing phacoemulsification at our institution; and 2) laboratory investigation of the effect of metformin on the behavior of lens epithelial cells in the context of an animal model for PCO. Population-averaged Cox proportional hazards modeling was used to estimate risk for time to Nd:YAG. For laboratory studies, expression of markers for epithelial-to-mesenchymal transition (EMT) implicated in PCO pathogenesis was measured in tissue culture and following extracapsular lens extraction in a mouse model. Results: The rate of Nd:YAG laser capsulotomy was 13.1% among the 9798 eyes. Both metformin use and diabetes were protective factors for Nd:YAG laser capsulotomy in univariate analysis. However, in multivariable analysis with nondiabetics as the reference group, only metformin use among diabetics was significantly protective of Nd:YAG (hazard ratio: 0.68, 95% CI: 0.54-0.85, P = 0.0008), while eyes of patients with diabetes without metformin use did not significantly differ (P = 0.5026). Treatment of lens epithelial cells with metformin reduced the level of the EMT markers ⍺-SMA and pERK induced by TGF-ß2. Similarly, metformin treatment reduced ⍺-SMA expression in lens epithelial cells following extracapsular lens extraction in a mouse model. Conclusions: The protective effect of metformin against early Nd:YAG may relate to its ability to downregulate EMT in residual lens epithelial cells that otherwise trend toward myofibroblast development and PCO.


Subject(s)
Capsule Opacification/therapy , Laser Therapy/methods , Lasers, Solid-State/therapeutic use , Metformin/therapeutic use , Posterior Capsule of the Lens/drug effects , Posterior Capsulotomy/methods , Postoperative Complications/prevention & control , Aged , Female , Follow-Up Studies , Humans , Hypoglycemic Agents/therapeutic use , Lenses, Intraocular , Male , Middle Aged , Posterior Capsule of the Lens/surgery , Retrospective Studies , Time Factors , Treatment Outcome
7.
Metabolites ; 11(7)2021 Jul 10.
Article in English | MEDLINE | ID: mdl-34357344

ABSTRACT

Aldose reductase (AR), the first and rate-limiting enzyme of the polyol pathway, has been implicated in the onset and development of the ocular complications of diabetes, including cataracts and retinopathy. Despite decades of research conducted to address possible mechanisms, questions still persist in understanding if or how AR contributes to imbalances leading to diabetic eye disease. To address these questions, we created a strain of transgenic mice engineered for the overexpression of human AR (AR-Tg). In the course of monitoring these animals for age-related retinal phenotypes, we observed signs of Müller cell gliosis characterized by strong immunostaining for glial fibrillary acidic protein. In addition, we observed increased staining for Iba1, consistent with an increase in the number of retinal microglia, a marker of retinal inflammation. Compared to age-matched nontransgenic controls, AR-Tg mice showed an age-dependent loss of Brn3a-positive retinal ganglion cells and an associated decrease in PERG amplitude. Both RGC-related phenotypes were rescued in animals treated with Sorbinil in drinking water. These results support the hypothesis that increased levels of AR may be a risk factor for structural and functional changes known to accompany retinopathy in humans.

8.
Chem Biol Interact ; 344: 109495, 2021 Aug 01.
Article in English | MEDLINE | ID: mdl-33961834

ABSTRACT

Cataracts, a clouding of the eye lens, are a leading cause of visual impairment and are responsible for one of the most commonly performed surgical procedures worldwide. Although generally safe and effective, cataract surgery can lead to a secondary lens abnormality due to transition of lens epithelial cells to a mesenchymal phenotype (EMT) and opacification of the posterior lens capsular bag. Occurring in up to 40% of cataract cases over time, posterior capsule opacification (PCO) introduces additional treatment costs and reduced quality of life for patients. Studies have shown that PCO pathogenesis is driven in part by TGF-ß, signaling through the action of the family of Smad coactivators to effect changes in gene transcription. In the present study, we evaluated the ability of Smad-7, a well characterized inhibitor of TGF-ß -mediated Smad signaling, to suppress the EMT response in lens epithelial cells associated with PCO pathogenesis. Treatment of lens epithelial cells with a cell-permeable form of Smad7 variant resulted in suppressed expression of EMT markers such as alpha smooth muscle actin and fibronectin. A single application of cell-permeable Smad7 variant in the capsular bag of a mouse cataract surgery model resulted in suppression of gene transcripts encoding alpha smooth muscle actin and fibronectin. These results point to Smad7 as a promising biotherapeutic agent for prevention or substantial reduction in the incidence of PCO following cataract surgery.


Subject(s)
Capsule Opacification/prevention & control , Cell-Penetrating Peptides/therapeutic use , Epithelial-Mesenchymal Transition/drug effects , Gene Products, tat/therapeutic use , Lens, Crystalline/drug effects , Smad7 Protein/therapeutic use , Actins/metabolism , Animals , Capsule Opacification/etiology , Capsule Opacification/pathology , Cataract/complications , Cataract/pathology , Epithelial Cells/drug effects , Lens, Crystalline/pathology , Mice, Transgenic , Protein Domains , Recombinant Proteins/therapeutic use
9.
Chem Biol Interact ; 307: 58-62, 2019 Jul 01.
Article in English | MEDLINE | ID: mdl-31026421

ABSTRACT

After cataract surgery, epithelial cells lining the anterior lens capsule can transition to one of two divergent pathways, including fibrosis which leads to posterior capsular opacification (PCO), or lens fiber cell differentiation which leads to regeneration of lens material. We previously showed that the PCO response can be suppressed with aldose reductase (AR) inhibitors. In this present study we show that AR inhibition, both genetic and pharmacologic with Sorbinil, can augment the process of lens regeneration. Extracapsular lens extraction (ECLE) was carried out in C57BL/6 (WT), AR overexpression (AR-Tg), and AR knockout (ARKO) mice, and in some cases in mice treated with the AR inhibitor sorbinil. Whole eyes were harvested approximately 8 weeks after ECLE and evaluated by histological analysis and immunostaining for the fiber cell marker γ-crystallin. All eyes examined for lens regeneration were paraffin embedded for serial sectioning to produce three-dimensional reconstructed models of lens morphology and size. We observed that AR-null mice respond to ECLE by regenerating a lens-like structure with a circular shape and array of cell nuclei reminiscent of the lens bow region typical of the native mammalian lens. Although WT and AR-Tg eyes also produced some regenerated lens material after ECLE, their structures were consistently smaller than ARKO regenerated lenses. WT mice treated with sorbinil showed higher levels of lens regeneration after ECLE compared to WT mice, as assessed by size and three-dimensional morphology. Altogether, this study adds evidence for a critical role for AR in the response of lens epithelial cells to cataract extraction and lens regeneration.


Subject(s)
Aldehyde Reductase/metabolism , Lens, Crystalline/physiology , Regeneration , Aldehyde Reductase/antagonists & inhibitors , Aldehyde Reductase/genetics , Animals , Cataract Extraction , Enzyme Inhibitors/pharmacology , Eye/diagnostic imaging , Imaging, Three-Dimensional , Imidazolidines/pharmacology , Lens, Crystalline/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Regeneration/drug effects
10.
Arterioscler Thromb Vasc Biol ; 38(11): 2691-2705, 2018 11.
Article in English | MEDLINE | ID: mdl-30354230

ABSTRACT

Objective- Blood-CNS (central nervous system) barrier defects are implicated in retinopathies, neurodegenerative diseases, stroke, and epilepsy, yet, the pathological mechanisms downstream of barrier defects remain incompletely understood. Blood-retina barrier (BRB) formation and retinal angiogenesis require ß-catenin signaling induced by the ligand norrin (NDP [Norrie disease protein]), the receptor FZD4 (frizzled 4), coreceptor LRP5 (low-density lipoprotein receptor-like protein 5), and the tetraspanin TSPAN12 (tetraspanin 12). Impaired NDP/FZD4 signaling causes familial exudative vitreoretinopathy, which may lead to blindness. This study seeked to define cell type-specific functions of TSPAN12 in the retina. Approach and Results- A loxP-flanked Tspan12 allele was generated and recombined in endothelial cells using a tamoxifen-inducible Cdh5-CreERT2 driver. Resulting phenotypes were documented using confocal microscopy. RNA-Seq, histopathologic analysis, and electroretinogram were performed on retinas of aged mice. We show that TSPAN12 functions in endothelial cells to promote vascular morphogenesis and BRB formation in developing mice and BRB maintenance in adult mice. Early loss of TSPAN12 in endothelial cells causes lack of intraretinal capillaries and increased VE-cadherin (CDH5 [cadherin5 aka VE-cadherin]) expression, consistent with premature vascular quiescence. Late loss of TSPAN12 strongly impairs BRB maintenance without affecting vascular morphogenesis, pericyte coverage, or perfusion. Long-term BRB defects are associated with immunoglobulin extravasation, complement deposition, cystoid edema, and impaired b-wave in electroretinograms. RNA-sequencing reveals transcriptional responses to the perturbation of the BRB, including genes involved in vascular basement membrane alterations in diabetic retinopathy. Conclusions- This study establishes mice with late endothelial cell-specific loss of Tspan12 as a model to study pathological consequences of BRB impairment in an otherwise intact vasculature.


Subject(s)
Blood-Retinal Barrier/metabolism , Endothelial Cells/metabolism , Retinal Neovascularization , Retinal Vessels/metabolism , Tetraspanins/deficiency , Age Factors , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Basement Membrane/metabolism , Basement Membrane/pathology , Blood-Retinal Barrier/immunology , Blood-Retinal Barrier/pathology , Cadherins/genetics , Cadherins/metabolism , Cell Proliferation , Cellular Senescence , Complement System Proteins/immunology , Complement System Proteins/metabolism , Diabetic Retinopathy/genetics , Diabetic Retinopathy/metabolism , Diabetic Retinopathy/pathology , Endothelial Cells/immunology , Endothelial Cells/pathology , Eye Diseases, Hereditary/genetics , Eye Diseases, Hereditary/metabolism , Eye Diseases, Hereditary/pathology , Familial Exudative Vitreoretinopathies , Female , Genotype , Immunoglobulins/immunology , Immunoglobulins/metabolism , Macular Edema/genetics , Macular Edema/metabolism , Macular Edema/pathology , Male , Mice, Knockout , Phenotype , Retinal Diseases/genetics , Retinal Diseases/metabolism , Retinal Diseases/pathology , Retinal Vessels/immunology , Retinal Vessels/pathology , Signal Transduction , Tetraspanins/genetics
11.
Invest Ophthalmol Vis Sci ; 59(8): 3591-3598, 2018 07 02.
Article in English | MEDLINE | ID: mdl-30025084

ABSTRACT

Purpose: Cataract surgery is a procedure by which the lens fiber cell mass is removed from its capsular bag and replaced with a synthetic intraocular lens. Postoperatively, remnant lens epithelial cells can undergo an aberrant wound healing response characterized by an epithelial-to-mesenchymal transition (EMT), leading to posterior capsular opacification (PCO). Aldose reductase (AR) inhibition has been shown to decrease EMT markers in cell culture models. In this study, we aim to demonstrate that AR inhibition can attenuate induction of EMT markers in an in vivo model of cataract surgery. Methods: A modified extracapsular lens extraction (ECLE) was performed on C57BL/6 wildtype, AR overexpression (AR-Tg), and AR knockout mice. Immunofluorescent staining for the myofibroblast marker α-smooth muscle actin (α-SMA), epithelial marker E-cadherin, and lens fiber cell markers αA-crystallin and Aquaporin 0 was used to characterize postoperative PCO. Quantitative reverse transcription PCR (qRT-PCR) was employed to quantify postoperative changes in α-SMA, vimentin, fibronectin, and E-cadherin. In a separate experiment, the AR inhibitor Sorbinil was applied postoperatively and qRT-PCR was used to assess changes in EMT markers. Results: Genetic AR knockout reduced ECLE-induced upregulation of α-SMA and downregulation of E-cadherin. These immunofluorescent changes were mirrored quantitatively in changes in mRNA levels. Similarly, Sorbinil blocked characteristic postoperative EMT changes in AR-Tg mice. Interestingly, genetic AR knockout did not prevent postoperative induction of the lens fiber cell markers αA-crystallin and Aquaporin 0. Conclusions: AR inhibition prevents the postoperative changes in EMT markers characteristic of PCO yet preserves the postoperative induction of lens fiber cell markers.


Subject(s)
Aldehyde Reductase/antagonists & inhibitors , Capsule Opacification/prevention & control , Cataract Extraction/adverse effects , Enzyme Inhibitors/pharmacology , Lens, Crystalline/pathology , Actins/biosynthesis , Actins/genetics , Animals , Cadherins/metabolism , Capsule Opacification/genetics , Capsule Opacification/pathology , Cell Movement , Cell Proliferation , Cells, Cultured , Disease Models, Animal , Epithelial-Mesenchymal Transition/drug effects , Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation , Lens, Crystalline/enzymology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction
12.
J AAPOS ; 22(1): 22-26.e1, 2018 02.
Article in English | MEDLINE | ID: mdl-29225157

ABSTRACT

BACKGROUND: Amniotic membrane grafts (AMGs) are used, with mixed results, as a platform for ocular healing and to reduce pathologic scarring. This study evaluated wound tensile strength and histopathologic changes after strabismus surgery with AMGs in 20 New Zealand white rabbits. METHODS: All subjects underwent 4 mm inferior rectus hang-back recessions to both eyes. The right eyes served as controls. Ten left eyes (group 1) received processed dehydrated amniotic membrane allografts (Ambiodry2, IOP Inc, Costa Mesa, CA) and ten left eyes (group 2) received cryopreserved human amniotic membrane allografts (AmnioGraft, Bio-Tissue, Miami, FL) between the sclera and muscle insertion and between the muscle and repositioned conjunctiva. At postoperative month 1, tensile strengths of the muscle-globe and conjunctiva-globe attachments were measured, and histopathologic analysis of each eye was performed. RESULTS: In group 1 the mean tensile strength of the muscle-globe attachments was 441.4 ± 274.4 g; of the conjunctiva-globe attachments, 640.3 ± 266.4 g. In the control eyes, the comparable values were 365.8 ± 199.8 g and 595.2 ± 315.3 g, respectively (P = 0.19, P = 0.13). In group 2 the mean tensile strengths were 456 ± 297.5 g and 608.2 ± 306.7 g, compared with control values of 352.7 ± 114.8 g and 583.8 ± 347.1 g (P = 0.43, P = 0.45). CONCLUSIONS: There was no significant change in tensile strength of the muscle insertion using AMGs. In a rabbit model, AMGs do not reduce inflammation or improve scar formation 1 month after strabismus surgery.


Subject(s)
Amnion/transplantation , Oculomotor Muscles/surgery , Ophthalmologic Surgical Procedures/methods , Strabismus/surgery , Tensile Strength/physiology , Wound Healing/physiology , Animals , Conjunctiva/pathology , Disease Models, Animal , Fibrosis/prevention & control , Humans , Postoperative Complications/prevention & control , Rabbits , Tissue Adhesions/pathology , Tissue Adhesions/prevention & control
13.
Mol Vis ; 22: 1095-1102, 2016.
Article in English | MEDLINE | ID: mdl-27667918

ABSTRACT

PURPOSE: Dry eye syndrome (DES) affects millions of people worldwide. Homeopathic remedies to treat a wide variety of ocular diseases have previously been documented in the literature, but little systematic work has been performed to validate the remedies' efficacy using accepted laboratory models of disease. The purpose of this study was to evaluate the efficacy of human milk and nopal cactus (prickly pear), two widely used homeopathic remedies, as agents to reduce pathological markers of DES. METHODS: The previously described benzalkonium chloride (BAK) dry eye mouse model was used to study the efficacy of human milk and nopal cactus (prickly pear). BAK (0.2%) was applied to the mouse ocular surface twice daily to induce dry eye pathology. Fluorescein staining was used to verify that the animals had characteristic signs of DES. After induction of DES, the animals were treated with human milk (whole and fat-reduced), nopal, nopal extract derivatives, or cyclosporine four times daily for 7 days. Punctate staining and preservation of corneal epithelial thickness, measured histologically at the end of treatment, were used as indices of therapeutic efficacy. RESULTS: Treatment with BAK reduced the mean corneal epithelial thickness from 36.77±0.64 µm in the control mice to 21.29±3.2 µm. Reduction in corneal epithelial thickness was largely prevented by administration of whole milk (33.2±2.5 µm) or fat-reduced milk (36.1±1.58 µm), outcomes that were similar to treatment with cyclosporine (38.52±2.47 µm), a standard in current dry eye therapy. In contrast, crude or filtered nopal extracts were ineffective at preventing BAK-induced loss of corneal epithelial thickness (24.76±1.78 µm and 27.99±2.75 µm, respectively), as were solvents used in the extraction of nopal materials (26.53±1.46 µm for ethyl acetate, 21.59±5.87 µm for methanol). Epithelial damage, as reflected in the punctate scores, decreased over 4 days of treatment with whole and fat-reduced milk but continued to increase in eyes treated with nopal-derived materials. CONCLUSIONS: Whole and fat-reduced human milk showed promising effects in the prevention of BAK-induced loss of corneal epithelial thickness and epithelial damage in this mouse model. Further studies are required to determine whether human milk may be safely used to treat dry eye in patients.

14.
J Spec Oper Med ; 16(4): 54-58, 2016.
Article in English | MEDLINE | ID: mdl-28088818

ABSTRACT

BACKGROUND: Success in Special Operations Forces medicine (SOFMED) depends on maximizing visual capability without compromising the provider or casualty when under fire. There is no single light that has been deemed "ideal" for all SOFMED environments. METHODS: We used the Farnsworth-Munsell (FM) hue test to determine color vision of normal subjects under white, red-green, and blue flashlights to determine color discrimination. Then we used a timed color-determination visual test to determine how quickly normal subjects can identify color correctly. We had subjects perform a simulated surgery illuminated by a normal white-light source, then by red-green or blue light-emitting diode (LED) tactical light. RESULTS: The total error score for white light was 49.714, 272.923 for red/green light, and 531.4 for blue light. The subjective perception of simulated trauma wounds was not substantially different with red-green LED tactical light when compared with white LED light. However, simulated surgery under the blue LED was more difficult compared with simulated surgery under the red-green LED light. CONCLUSION: Red-green was a superior light source for SOFMED and military first responders in this study, especially, where light was required to allow accurate and efficient application of Tactical Combat Casualty Care to injured personnel.


Subject(s)
Color Vision , Color , Lighting , Military Medicine , Surgical Procedures, Operative , Adolescent , Adult , Aged , Female , Humans , Male , Middle Aged , Vision, Ocular , Young Adult
15.
PLoS One ; 10(9): e0137659, 2015.
Article in English | MEDLINE | ID: mdl-26355842

ABSTRACT

Misfolded protein aggregation, including cataract, cause a significant amount of blindness worldwide. α-Crystallin is reported to bind misfolded proteins and prevent their aggregation. We hypothesize that supplementing retina and lens with α-crystallin may help to delay disease onset. The purpose of this study was to determine if αB-crystallin subunits containing a cell penetration peptide (gC-tagged αB-crystallin) facilitate the uptake of wild type αA-crystallin (WT-αA) in lens and retina. Recombinant human αB-crystallin was modified by the addition of a novel cell penetration peptide derived from the gC gene product of herpes simplex virus (gC-αB). Recombinant gC-αB and wild-type αA-crystallin (WT-αA) were purified from E. coli over-expression cultures. After Alexa-labeling of WT-αA, these proteins were mixed at ratios of 1:2, 1:5 and 1:10, respectively, and incubated at 37°C for 4 hours to allow for subunit exchange. Mixed oligomers were subsequently incubated with tissue culture cells or mouse organ cultures. Similarly, crystallin mixtures were injected into the vitreous of rat eyes. At various times after exposure, tissues were harvested and analyzed for protein uptake by confocal microscopy or flow cytometry. Chaperone-like activity assays were performed on α-crystallins ratios showing optimal uptake using chemically-induced or heat induced substrate aggregation assays. As determined by flow cytometry, a ratio of 1:5 for gC-αB to WT-αA was found to be optimal for uptake into retinal pigmented epithelial cells (ARPE-19). Chaperone-like activity assays demonstrated that hetero-oligomeric complex of gC-αB to WT-αA (in 1:5 ratio) retained protein aggregation protection. We observed a significant increase in protein uptake when optimized (gC-αB to WT-αA (1:5 ratio)) hetero-oligomers were used in mouse lens and retinal organ cultures. Increased levels of α-crystallin were found in lens and retina following intravitreal injection of homo- and hetero-oligomers in rats.


Subject(s)
Lens, Crystalline/metabolism , Retina/metabolism , alpha-Crystallins/metabolism , Animals , Epithelial Cells/metabolism , Humans , Mice , Protein Multimerization , Rats , Retinal Pigment Epithelium/metabolism , alpha-Crystallin A Chain/chemistry , alpha-Crystallin A Chain/metabolism , alpha-Crystallin B Chain/chemistry , alpha-Crystallin B Chain/metabolism , alpha-Crystallins/chemistry
16.
Biochim Biophys Acta ; 1843(2): 309-15, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24275510

ABSTRACT

In addition to their key role as structural lens proteins, α-crystallins also appear to confer protection against many eye diseases, including cataract, retinitis pigmentosa, and macular degeneration. Exogenous recombinant α-crystallin proteins were examined for their ability to prevent cell death induced by heat or oxidative stress in a human lens epithelial cell line (HLE-B3). Wild type αA- or αB-crystallin (WT-αA and WT-αB) and αA- or αB-crystallins, modified by the addition of a cell penetration peptide (CPP) designed to enhance the uptake of proteins into cells (gC-αB, TAT-αB, gC-αA), were produced by recombinant methods. In vitro chaperone-like assays were used to assay the ability of α-crystallins to protect client proteins from chemical or heat induced aggregation. In vivo viability assays were performed in HLE-B3 to determine whether pre-treatment with α-crystallins reduced death after exposure to oxidative or heat stress. Most of the five recombinant α-crystallin proteins tested conferred some in vitro protection from protein aggregation, with the greatest effect seen with WT-αB and gC-αB. All α-crystallins displayed significant protection to oxidative stress induced cell death, while only the αB-crystallins reduced cell death induced by thermal stress. Our findings indicate that the addition of the gC tag enhanced the protective effect of αB-crystallin against oxidative but not thermally-induced cell death. In conclusion, modifications that increase the uptake of α-crystallin proteins into cells, without destroying their chaperone-like activity and anti-apoptotic functions, create the potential to use these proteins therapeutically.


Subject(s)
Crystallins/pharmacology , Cytoprotection/drug effects , Hot Temperature , Lens, Crystalline/pathology , Oxidative Stress/drug effects , alpha-Crystallin B Chain/pharmacology , Aldehyde Reductase/metabolism , Cell Death/drug effects , Cell Line , Cell Survival/drug effects , Cell-Penetrating Peptides/pharmacology , Crystallins/metabolism , Humans , Protein Structure, Quaternary , Recombinant Proteins/pharmacology , Recombinant Proteins/toxicity
17.
Cytokine ; 29(4): 176-85, 2005 Feb 21.
Article in English | MEDLINE | ID: mdl-15652450

ABSTRACT

OBJECTIVES: In pregnancy, the placental contribution of cytokines to maternal immunosuppression has been established, however their role in normal maternal blood pressure regulation has not been identified. We investigate the contribution of interleukin-10 (IL-10) and tumor necrosis factor-alpha (TNF-alpha) to the vasodilation of early pregnancy in non-human primates. We also sequenced the IL-10 baboon gene and compared it with humans. METHODS: The effect of four different treatments, administered sequentially (semi-random-design) on resting 18h, night time, or hourly mean arterial pressure (MAP) and heart rate (HR) were measured using telemetry. An anti-human IL-10 monoclonal antibody (MAb, 1mg, n=7), anti-TNF-alpha antibody (n=3), a combination of anti-IL-10 and anti-TNF-alpha antibodies (n=5) or saline (n=3) control were administered intravenously to baboons in early pregnancy. Plasma and placental IL-10 concentration was measured before and after injection in all animals. RESULTS: Anti-human IL-10 MAb caused a significant increase in MAP of 2.6+/-0.5mmHg over the 18-h period (p<0.05). Administration of TNF-alpha alone or in combination with IL-10 did not alter MAP. There was 97% sequence homology of IL-10 cDNA between humans and baboons. CONCLUSIONS: IL-10 was shown to regulate the vasodilation of early pregnancy in Papio hamadryas. This partial role of IL-10 in the early BP response of primate pregnancy may be relevant to pathophysiological states of human pregnancy such as preeclampsia.


Subject(s)
Blood Pressure/physiology , Interleukin-10/physiology , Amino Acid Sequence/genetics , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Blood Pressure/drug effects , Cells, Cultured , Creatinine/urine , Endometrium/cytology , Endometrium/drug effects , Endometrium/metabolism , Female , Gene Expression/drug effects , Interleukin-10/genetics , Interleukin-10/immunology , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/metabolism , Molecular Sequence Data , Papio hamadryas , Phytohemagglutinins/pharmacology , Placenta/drug effects , Placenta/metabolism , Pregnancy , Proteinuria/urine , Sequence Homology, Amino Acid , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/physiology
18.
Cancer Gene Ther ; 10(10): 745-54, 2003 Oct.
Article in English | MEDLINE | ID: mdl-14502227

ABSTRACT

We have developed a novel dual-fluorescence reporter system incorporating green (GFP) and red (RFP) fluorescent proteins to monitor expression of the N-ras(m) gene and an N-ras(m) suppressor, respectively. Retroviral vectors were produced in which human N-ras(m) (codon 13 mutation) was coexpressed with GFP, and a ribozyme specifically targeting N-ras(m) was coexpressed with RFP. N-Ras(m) suppression was monitored by measurement of GFP fluorescence in dual-fluorescent (GFP and RFP) cells. We demonstrated that the degree of N-ras(m) suppression was dependent on the ribozyme dose, proportional to red fluorescence, in dual-fluorescent cells. We further showed that ribozyme-mediated N-ras(m)suppression inhibited growth of NIH3T3 and CD34-positive TF-1 cells. In these cultures, ras suppressor activity resulted in the depletion of suppressor-positive cells due to inhibition of cell growth. In contrast, N-ras(m) suppression produced a growth advantage to human leukemic K562 cells, presumably by inhibiting N-ras(m)-induced apoptosis. In K562 cells, ras suppression resulted in the outgrowth of suppressor-positive cells. This provides a platform to identify suppressors of ras that is based on function.


Subject(s)
Apoptosis , Genes, Reporter/genetics , RNA, Catalytic/metabolism , ras Proteins/antagonists & inhibitors , ras Proteins/genetics , Animals , Butadienes/pharmacology , Cell Line, Tumor , Chromones/pharmacology , Flow Cytometry , Fluorescence , Genetic Vectors/genetics , Humans , K562 Cells , MAP Kinase Signaling System , Mice , Morpholines/pharmacology , Mutation/genetics , NIH 3T3 Cells , Nitriles/pharmacology , RNA, Catalytic/genetics , ras Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...